Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 243
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 121(18): e2317760121, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38652741

ABSTRACT

The complex interplay between malignant cells and the cellular and molecular components of the tumor stroma is a key aspect of cancer growth and development. These tumor-host interactions are often affected by soluble bioactive molecules such as proteoglycans. Decorin, an archetypical small leucine-rich proteoglycan primarily expressed by stromal cells, affects cancer growth in its soluble form by interacting with several receptor tyrosine kinases (RTK). Overall, decorin leads to a context-dependent and protracted cessation of oncogenic RTK activity by attenuating their ability to drive a prosurvival program and to sustain a proangiogenic network. Through an unbiased transcriptomic analysis using deep RNAseq, we identified that decorin down-regulated a cluster of tumor-associated genes involved in lymphatic vessel (LV) development when systemically delivered to mice harboring breast carcinoma allografts. We found that Lyve1 and Podoplanin, two established markers of LVs, were markedly suppressed at both the mRNA and protein levels, and this suppression correlated with a significant reduction in tumor LVs. We further identified that soluble decorin, but not its homologous proteoglycan biglycan, inhibited LV sprouting in an ex vivo 3D model of lymphangiogenesis. Mechanistically, we found that decorin interacted with vascular endothelial growth factor receptor 3 (VEGFR3), the main lymphatic RTK, and its activity was required for the decorin-mediated block of lymphangiogenesis. Finally, we identified that Lyve1 was in part degraded via decorin-evoked autophagy in a nutrient- and energy-independent manner. These findings implicate decorin as a biological factor with antilymphangiogenic activity and provide a potential therapeutic agent for curtailing breast cancer growth and metastasis.


Subject(s)
Decorin , Lymphangiogenesis , Decorin/metabolism , Decorin/genetics , Animals , Mice , Humans , Female , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Lymphatic Vessels/metabolism , Lymphatic Vessels/pathology , Cell Line, Tumor , Disease Progression , Vesicular Transport Proteins/metabolism , Vesicular Transport Proteins/genetics , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Gene Expression Regulation, Neoplastic
2.
Am J Physiol Cell Physiol ; 325(2): C391-C405, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37399497

ABSTRACT

Progranulin is a growth factor with pro-tumorigenic activity. We recently demonstrated that in mesothelioma, progranulin regulates cell migration, invasion, adhesion, and in vivo tumor formation by modulating a complex signaling network involving multiple receptor tyrosine kinase (RTK)s. Progranulin biological activity relies on epidermal growth factor receptor (EGFR) and receptor-like tyrosine kinase (RYK), a co-receptor of the Wnt signaling pathway, which are both required for progranulin-induced downstream signaling. However, the molecular mechanism regulating the functional interaction among progranulin, EGFR, and RYK are not known. In this study, we demonstrated that progranulin directly interacted with RYK by specific enzyme-linked immunosorbent assay (ELISA) (KD = 0.67). Using immunofluorescence and proximity ligation assay, we further discovered that progranulin and RYK colocalized in mesothelioma cells in distinct vesicular compartments. Notably, progranulin-dependent downstream signaling was sensitive to endocytosis inhibitors, suggesting that it could depend on RYK or EGFR internalization. We discovered that progranulin promoted RYK ubiquitination and endocytosis preferentially through caveolin-1-enriched pathways, and modulated RYK stability. Interestingly, we also showed that in mesothelioma cells, RYK complexes with the EGFR, contributing to the regulation of RYK stability. Collectively, our results suggest a complex regulation of RYK trafficking/activity in mesothelioma cells, a process that is concurrently regulated by exogenous soluble progranulin and EGFR. NEW & NOTEWORTHY The growth factor progranulin has pro-tumorigenic activity. In mesothelioma, progranulin signaling is mediated by EGFR and RYK, a co-receptor of the Wnt signaling. However, the molecular mechanisms regulating progranulin action are not well defined. Here, we demonstrated that progranulin binds RYK and regulates its ubiquitination, internalization, and trafficking. We also uncovered a role for EGFR in modulating RYK stability. Overall, these results highlight a complex modulation of RYK activity by progranulin and EGFR in mesothelioma.


Subject(s)
Mesothelioma , Receptor Protein-Tyrosine Kinases , Humans , Progranulins , Receptor Protein-Tyrosine Kinases/metabolism , ErbB Receptors/metabolism , Wnt Signaling Pathway/physiology , Cell Movement , Intercellular Signaling Peptides and Proteins
3.
Am J Physiol Cell Physiol ; 323(5): C1355-C1373, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36036446

ABSTRACT

Decorin, a small leucine-rich proteoglycan with multiple biological functions, is known to evoke autophagy and mitophagy in both endothelial and cancer cells. Here, we investigated the effects of soluble decorin on mitochondrial homeostasis using live cell imaging and ex vivo angiogenic assays. We discovered that decorin triggers mitochondrial depolarization in triple-negative breast carcinoma, HeLa, and endothelial cells. This bioactivity was mediated by the protein core in a time- and dose-dependent manner and was specific for decorin insofar as biglycan, the closest homolog, failed to trigger depolarization. Mechanistically, we found that the bioactivity of decorin to promote depolarization required the MET receptor and its tyrosine kinase. Moreover, two mitochondrial interacting proteins, mitostatin and mitofusin 2, were essential for downstream decorin effects. Finally, we found that decorin relied on the canonical mitochondrial permeability transition pore to trigger tumor cell mitochondrial depolarization. Collectively, our study implicates decorin as a soluble outside-in regulator of mitochondrial dynamics.


Subject(s)
Carcinoma , Decorin , Endothelial Cells , Humans , Biglycan/pharmacology , Carcinoma/metabolism , Decorin/pharmacology , Endothelial Cells/metabolism , Extracellular Matrix Proteins/metabolism , Mitochondrial Permeability Transition Pore , Protein-Tyrosine Kinases/metabolism , Signal Transduction
4.
Am J Physiol Cell Physiol ; 322(3): C554-C566, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35171698

ABSTRACT

Decorin is a stromal-derived prototype member of the small leucine-rich proteoglycan gene family. In addition to its functions as a regulator of collagen fibrillogenesis and TGF-ß activity soluble decorin acts as a pan-receptor tyrosine kinase (RTK) inhibitor. Decorin binds to various RTKs including EGFR HER2 HGFR/Met VEGFR2 TLR and IGFR. Although the molecular mechanism for the action of decorin on these receptors is not entirely elucidated overall decorin evokes transient activation of these receptors with suppression of downstream signaling cascades culminating in growth inhibition followed by their physical downregulation via caveosomal internalization and degradation. In the case of Met decorin leads to decreased ß-catenin signaling pathway and growth suppression. As most of these RTKs are responsible for providing a growth advantage to cancer cells the result of decorin treatment is oncosuppression. Another decorin-driven mechanism to restrict cancer growth and dissemination is by impeding angiogenesis via vascular endothelial growth factor receptor 2 (VEGFR2) and the concurrent activation of protracted endothelial cell autophagy. In this review we will dissect the multiple roles of decorin in cancer biology and its potential use as a next-generation protein-based adjuvant therapy to combat cancer.


Subject(s)
Neoplasms , Vascular Endothelial Growth Factor A , Autophagy/physiology , Carrier Proteins/metabolism , Decorin/genetics , Decorin/metabolism , Extracellular Matrix Proteins/metabolism , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism
5.
Exp Eye Res ; 216: 108933, 2022 03.
Article in English | MEDLINE | ID: mdl-35031282

ABSTRACT

A characteristic rigid spatial arrangement of collagen fibrils in the stroma is critical for corneal transparency. This unique organization of collagen fibrils in corneal stroma can be impacted by the presence and interactions of proteoglycans and extracellular matrix (ECM) proteins in a corneal microenvironment. Earlier studies revealed that decorin, a leucine-rich proteoglycan in stroma, regulates keratocyte-collagen matrix assembly and wound healing in the cornea. This study investigated the role of decorin in the regulation of stromal fibrillogenesis and corneal transparency in vivo employing a loss-of-function genetic approach using decorin null (dcn-/-) and wild type (dcn+/+) mice and a standard alkali-injury model. A time-dependent ocular examinations with Slit lamp microscope in live animals assessed corneal clarity, haze, and neovascularization levels in normal and injured eyes. Morphometric changes in normal and injured dcn+/+ and dcn-/- corneas, post-euthanasia, were analyzed with Masson's Trichrome and Periodic Acid-Schiff (PAS) histology evaluations. The ultrastructure changes in all corneas were investigated with transmission electron microscopy (TEM). Injury to eye produced clinically relevant corneal haze and neovascularization in dcn-/- and dcn+/+ mice while corneas of uninjured eyes remained clear and avascular. A clinically significant haze and neovascularization appeared in injured dcn-/- corneas compared to the dcn+/+ corneas at day 21 post-injury and not at early tested times. Histological examinations revealed noticeably abnormal morphology and compromised collagen levels in injured dcn-/- corneas compared to the injured/normal dcn+/+ and uninjured dcn-/- corneas. TEM analysis exhibited remarkably uneven collagen fibrils size and distribution in the stroma with asymmetrical organization and loose packing in injured dcn-/- corneas than injured/normal dcn+/+ and uninjured dcn-/- corneas. The minimum and maximum inter-fibril distances were markedly irregular in injured dcn-/- corneas compared to all other corneas. Together, results of clinical, histological, and ultrastructural investigations in a genetic knockout model suggested that decorin influenced stromal fibrillogenesis and transparency in healing cornea.


Subject(s)
Corneal Injuries/metabolism , Decorin/physiology , Fibrillar Collagens/metabolism , Organogenesis/physiology , Wound Healing/physiology , Animals , Burns, Chemical/metabolism , Corneal Injuries/pathology , Extracellular Matrix Proteins/metabolism , Eye Burns/chemically induced , Fibrillar Collagens/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Electron, Transmission , Slit Lamp Microscopy , Sodium Hydroxide
6.
J Biomech Eng ; 144(6)2022 06 01.
Article in English | MEDLINE | ID: mdl-35348624

ABSTRACT

Cervical remodeling is critical for a healthy pregnancy. The proper regulation of extracellular matrix (ECM) turnover leads to remodeling throughout gestation, transforming the tissue from a stiff material to a compliant, extensible, viscoelastic tissue prepared for delivery. Small leucine-rich proteoglycans (SLRPs) regulate structural fiber assembly in the cervical ECM and overall tissue material properties. To quantify the SLRPs' mechanical role in the cervix, whole cervix specimens from nonpregnant and late pregnant knockout mice of SLRPs, decorin and biglycan, were subjected to cyclic load-unload, ramp-hold, and load-to-failure mechanical tests. Further, a fiber composite material model, accounting for collagen fiber bundle waviness, was developed to describe the cervix's three-dimensional large deformation equilibrium behavior. In nonpregnant tissue, SLRP knockout cervices have the same equilibrium material properties as wild-type tissue. In contrast, the load-to-failure and ramp-hold tests reveal SLRPs impact rupture and time-dependent relaxation behavior. Loss of decorin in nonpregnant (NP) cervices results in inferior rupture properties. After extensive remodeling, cervical strength is similar between all genotypes, but the SLRP-deficient tissue has a diminished ability to dissipate stress during a ramp-hold. In mice with a combined loss of decorin and biglycan, the pregnant cervix loses its extensibility, compliance, and viscoelasticity. These results suggest that decorin and biglycan are necessary for crucial extensibility and viscoelastic material properties of a healthy, remodeled pregnant cervix.


Subject(s)
Cervix Uteri , Extracellular Matrix , Animals , Biglycan/genetics , Decorin/genetics , Extracellular Matrix Proteins/genetics , Female , Mice , Mice, Knockout , Pregnancy
7.
Semin Cancer Biol ; 62: 1-8, 2020 05.
Article in English | MEDLINE | ID: mdl-31078640

ABSTRACT

The need for more effective cancer therapies is omnipresent as the ever-complex, and highly adaptive, mechanisms of tumor biology allow this disease to elude even the most stringent treatment options. The expanding field of proteoglycan signaling is enticing as a reservoir of potential drug targets and prospects for novel therapeutic strategies. The newest trend in proteoglycan biology is the interplay between extracellular signaling and autophagy fueled by the close link between autophagy and angiogenesis. Here we summarize the most current evidence surrounding proteoglycan signaling in both of these biological processes featuring the well-known suspects, decorin and perlecan, as well as other up-and-coming neophytes in this evolving signaling web.


Subject(s)
Endothelial Cells/metabolism , Neovascularization, Pathologic/metabolism , Proteoglycans/metabolism , Signal Transduction , Animals , Autophagy , Biomarkers , Decorin/metabolism , Humans
8.
J Biol Chem ; 295(49): 16797-16812, 2020 12 04.
Article in English | MEDLINE | ID: mdl-33020183

ABSTRACT

The extracellular matrix encompasses a reservoir of bioactive macromolecules that modulates a cornucopia of biological functions. A prominent body of work posits matrix constituents as master regulators of autophagy and angiogenesis and provides molecular insight into how these two processes are coordinated. Here, we review current understanding of the molecular mechanisms underlying hyaluronan and HAS2 regulation and the role of soluble proteoglycan in affecting autophagy and angiogenesis. Specifically, we assess the role of proteoglycan-evoked autophagy in regulating angiogenesis via the HAS2-hyaluronan axis and ATG9A, a novel HAS2 binding partner. We discuss extracellular hyaluronan biology and the post-transcriptional and post-translational modifications that regulate its main synthesizer, HAS2. We highlight the emerging group of proteoglycans that utilize outside-in signaling to modulate autophagy and angiogenesis in cancer microenvironments and thoroughly review the most up-to-date understanding of endorepellin signaling in vascular endothelia, providing insight into the temporal complexities involved.


Subject(s)
Autophagy , Endothelium, Vascular/metabolism , Hyaluronic Acid/metabolism , Autophagy-Related Proteins/metabolism , Endothelium, Vascular/cytology , Humans , Hyaluronan Synthases/metabolism , Neovascularization, Pathologic , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
9.
J Biol Chem ; 295(19): 6344-6356, 2020 05 08.
Article in English | MEDLINE | ID: mdl-32205445

ABSTRACT

Endorepellin, the C-terminal fragment of the heparan sulfate proteoglycan perlecan, influences various signaling pathways in endothelial cells by binding to VEGFR2. In this study, we discovered that soluble endorepellin activates the canonical stress signaling pathway consisting of PERK, eIF2α, ATF4, and GADD45α. Specifically, endorepellin evoked transient activation of VEGFR2, which, in turn, phosphorylated PERK at Thr980 Subsequently, PERK phosphorylated eIF2α at Ser51, upregulating its downstream effector proteins ATF4 and GADD45α. RNAi-mediated knockdown of PERK or eIF2α abrogated the endorepellin-mediated up-regulation of GADD45α, the ultimate effector protein of this stress signaling cascade. To functionally validate these findings, we utilized an ex vivo model of angiogenesis. Exposure of the aortic rings embedded in 3D fibrillar collagen to recombinant endorepellin for 2-4 h activated PERK and induced GADD45α vis à vis vehicle-treated counterparts. Similar effects were obtained with the established cellular stress inducer tunicamycin. Notably, chronic exposure of aortic rings to endorepellin for 7-9 days markedly suppressed vessel sprouting, an angiostatic effect that was rescued by blocking PERK kinase activity. Our findings unravel a mechanism by which an extracellular matrix protein evokes stress signaling in endothelial cells, which leads to angiostasis.


Subject(s)
Aorta/metabolism , Heparan Sulfate Proteoglycans/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Peptide Fragments/metabolism , Signal Transduction , Stress, Physiological , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Animals , Aorta/cytology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Heparan Sulfate Proteoglycans/genetics , Human Umbilical Vein Endothelial Cells/cytology , Humans , Mice , Peptide Fragments/genetics , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
10.
J Biol Chem ; 295(18): 6064-6079, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32209654

ABSTRACT

Extracellular matrix-evoked angiostasis and autophagy within the tumor microenvironment represent two critical, but unconnected, functions of the small leucine-rich proteoglycan, decorin. Acting as a partial agonist of vascular endothelial growth factor 2 (VEGFR2), soluble decorin signals via the energy sensing protein, AMP-activated protein kinase (AMPK), in the autophagic degradation of intracellular vascular endothelial growth factor A (VEGFA). Here, we discovered that soluble decorin evokes intracellular catabolism of endothelial VEGFA that is mechanistically independent of mTOR, but requires an autophagic regulator, paternally expressed gene 3 (PEG3). We found that administration of autophagic inhibitors such as chloroquine or bafilomycin A1, or depletion of autophagy-related 5 (ATG5), results in accumulation of intracellular VEGFA, indicating that VEGFA is a basal autophagic substrate. Mechanistically, decorin increased the VEGFA clearance rate by augmenting autophagic flux, a process that required RAB24 member RAS oncogene family (RAB24), a small GTPase that facilitates the disposal of autophagic compartments. We validated these findings by demonstrating the physiological relevance of this process in vivo Mice starved for 48 h exhibited a sharp decrease in overall cardiac and aortic VEGFA that could be blocked by systemic chloroquine treatment. Thus, our findings reveal a unified mechanism for the metabolic control of endothelial VEGFA for autophagic clearance in response to decorin and canonical pro-autophagic stimuli. We posit that the VEGFR2/AMPK/PEG3 axis integrates the anti-angiogenic and pro-autophagic bioactivities of decorin as the molecular basis for tumorigenic suppression. These results support future therapeutic use of decorin as a next-generation protein therapy to combat cancer.


Subject(s)
Autophagy , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Proteolysis , Vascular Endothelial Growth Factor A/metabolism , AMP-Activated Protein Kinases/metabolism , Decorin/metabolism , Homeostasis , Humans , Intracellular Space/metabolism , Kruppel-Like Transcription Factors/metabolism , Nutrients/deficiency , rab GTP-Binding Proteins/metabolism
11.
Biol Reprod ; 104(3): 611-623, 2021 03 11.
Article in English | MEDLINE | ID: mdl-33165521

ABSTRACT

Approximately, 25% of all preterm births are due to preterm premature rupture of membranes. Mice deficient in proteoglycans biglycan (Bgn) and decorin (Dcn) display abnormal fetal membranes and increased incidence of preterm birth. We conducted RNA-Seq to profile fetal membranes and identify molecular pathways that may lead to preterm birth in double knockout (DKO) mice (Bgn-/-; Dcn-/-) compared to wild-type (WT) at two different gestational stages, E12 and E18 (n = 3 in each group). 3264 transcripts were differentially regulated in E18 DKO vs. WT fetal membranes, and 96 transcripts differentially regulated in E12 DKO vs. WT fetal membranes (FDR < 0.05, log 2 FC ≥ 1). Differentially regulated transcripts in E18 DKO fetal membranes were significantly enriched for genes involved in cell cycle regulation, extracellular matrix-receptor interaction, and the complement cascade. Fifty transcripts involved in the cell cycle were altered in E18 DKO fetal membranes (40↓, 10↑, FDR < 0.05), including p21 and p57 (↑), and Tgfb2, Smad3, CycA, Cdk1, and Cdk2(↓). Thirty-one transcripts involved in the complement cascade were altered (11↓, 20↑, FDR < 0.05) in E18 DKO fetal membranes, including C1q, C2, and C3 (↑). Differentially expressed genes in the top three molecular pathways (1) showed evidence of negative or purifying selection, and (2) were significantly enriched (Z-score > 10) for transcription factor binding sites for Nr2f1 at E18. We propose that in DKO mice, cell cycle arrest results in lack of cell proliferation in fetal membranes, inability to contain the growing fetus, and preterm birth.


Subject(s)
Biglycan/metabolism , Decorin/metabolism , Extraembryonic Membranes/metabolism , Gene Expression Regulation , Animals , Biglycan/genetics , Biological Evolution , Cell Cycle/physiology , Complement System Proteins/metabolism , Decorin/genetics , Disease Models, Animal , Extracellular Matrix , Humans , Infant, Newborn , Mice , Mice, Knockout , Premature Birth , RNA-Seq , Transcriptome
12.
Exp Eye Res ; 207: 108610, 2021 06.
Article in English | MEDLINE | ID: mdl-33940009

ABSTRACT

Our earlier decorin (Dcn) gene overexpression studies found that the targeted Dcn gene transfer into the cornea inhibited corneal angiogenesis in vivo using a rabbit model. In this study, we tested the hypothesis that anti-angiogenic effects of decorin in the cornea are mediated by alterations in a normal physiologic balance of pro- and anti-angiogenic factors using decorin deficient (Dcn-/-) and wild type (Dcn+/+) mice. Corneal neovascularization (CNV) in Dcn-/- and Dcn+/+ mice was produced with a standard chemical injury technique. The clinical progression of CNV in mice was monitored with stereo- and slit-lamp microscopes, and histopathological hematoxylin and eosin (H&E) staining. Protein and mRNA expression of pro- and anti-angiogenic factors in the cornea were evaluated using immunofluorescence and quantitative real-time PCR, respectively. Slit-lamp clinical eye examinations revealed significantly more CNV in Dcn-/- mice than the Dcn+/+ mice post-injury (p < 0.05) and AAV5-Dcn gene therapy significantly reduced CNV in Dcn-/- mice compered to no AAV5-Dcn gene therapy controls (p < 0.001). H&E-stained corneal sections exhibited morphology with several neovessels in injured corneas of the Dcn-/- mice than the Dcn+/+ mice. Immunofluorescence of corneal sections displayed significantly higher expression of α-smooth muscle actin (α-SMA) and endoglin proteins in Dcn-/- mice than Dcn+/+ mice (p < 0.05). Quantitative real-time PCR found significantly increased mRNA levels of pro-angiogenic factors endoglin (2.53-fold; p < 0.05), Vegf (2.47-fold; p < 0.05), and Pecam (2.14-fold; p < 0.05) and anti-angiogenic factor Vegfr2 (1.56-fold; p < 0.05) in the normal cornea of the Dcn-/- mice than the Dcn+/+ mice. Furthermore, neovascularized Dcn-/- mice corneas showed greater increase in mRNA expression of pro-angiogenic factors endoglin (4.58-fold; p < 0.0001), Vegf (4.16-fold; p < 0.0001), and Pdgf (2.15-fold; p < 0.0001) and reduced expression of anti-angiogenic factors Ang2 (0.12-fold; p < 0.05), Timp1 (0.22-fold; p < 0.05), and Vegfr2 (0.67-fold; p > 0.05) compared to neovascularized Dcn+/+ mice corneas. These gene deficience studies carried with transgenic Dcn-/- mice revealed decorin's role in influencing a physiologic balance between pro-and anti-angiogenic factors in the normal and injured cornea. We infer that the functional deletion of Dcn promotes irregular corneal repair and aggravates CNV.


Subject(s)
Corneal Neovascularization/metabolism , Corneal Neovascularization/physiopathology , Decorin/physiology , Actins/metabolism , Animals , Corneal Neovascularization/genetics , Endoglin/genetics , Endoglin/metabolism , Female , Gene Expression Regulation/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Platelet Endothelial Cell Adhesion Molecule-1/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics
13.
Int J Mol Sci ; 22(14)2021 Jul 17.
Article in English | MEDLINE | ID: mdl-34299278

ABSTRACT

During the pathogenesis of glaucoma, optic nerve (ON) axons become continuously damaged at the optic nerve head (ONH). This often is associated with reactive astrocytes and increased transforming growth factor (TGF-ß) 2 levels. In this study we tested the hypothesis if the presence or absence of decorin (DCN), a small leucine-rich proteoglycan and a natural inhibitor of several members of the TGF family, would affect the expression of the TGF-ßs and connective tissue growth factor (CTGF/CCN2) in human ONH astrocytes and murine ON astrocytes. We found that DCN is present in the mouse ON and is expressed by human ONH and murine ON astrocytes. DCN expression and synthesis was significantly reduced after 24 h treatment with 3 nM CTGF/CCN2, while treatment with 4 pM TGF-ß2 only reduced expression of DCN significantly. Conversely, DCN treatment significantly reduced the expression of TGF-ß1, TGF-ß2 and CTGF/CCN2 vis-a-vis untreated controls. Furthermore, DCN treatment significantly reduced expression of fibronectin (FN) and collagen IV (COL IV). Notably, combined treatment with DCN and triciribine, a small molecule inhibitor of protein kinase B (AKT), attenuated effects of DCN on CTGF/CCN2, TGF-ß1, and TGF-ß2 mRNA expression. We conclude (1) that DCN is an important regulator of TGF-ß and CTGF/CCN2 expression in astrocytes of the ON and ONH, (2) that DCN thereby regulates the expression of extracellular matrix (ECM) components and (3) that DCN executes its negative regulatory effects on TGF-ß and CTGF/CCN2 via the pAKT/AKT signaling pathway in ON astrocytes.


Subject(s)
Astrocytes/metabolism , Decorin/pharmacology , Extracellular Matrix Proteins/metabolism , Glaucoma/pathology , Oncogene Protein v-akt/metabolism , Optic Nerve/metabolism , Transforming Growth Factor beta2/antagonists & inhibitors , Animals , Astrocytes/drug effects , Cells, Cultured , Disease Models, Animal , Female , Glaucoma/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Optic Nerve/drug effects , Signal Transduction
14.
Chem Rev ; 118(18): 9152-9232, 2018 09 26.
Article in English | MEDLINE | ID: mdl-30204432

ABSTRACT

The extracellular matrix (ECM) constitutes a highly dynamic three-dimensional structural network comprised of macromolecules, such as proteoglycans/glycosaminoglycans (PGs/GAGs), collagens, laminins, fibronectin, elastin, other glycoproteins and proteinases. In recent years, the field of PGs has expanded rapidly. Due to their high structural complexity and heterogeneity, PGs mediate several homeostatic and pathological processes. PGs consist of a protein core and one or more covalently attached GAG chains, which provide the protein cores with the ability to interact with several proteins. The GAG building blocks of PGs significantly influence the chemical and functional properties of PGs. The primary goal of this comprehensive review is to summarize major achievements and paradigm-shifting discoveries made on the PG/GAG chemistry-biology axis, focusing on structural variability, structure-function relationships, metabolic, molecular, and epigenetic mechanisms underlying their synthesis. Recent insights related to exosome biogenesis, degradation, and cell signaling, their status as diagnostic tools and potential pharmacological targets in diseases as well as current applications in nanotechnology and biotechnology are addressed. Moreover, issues related to docking studies, molecular modeling, GAG/PG interaction networks, and their integration are discussed.


Subject(s)
Glycosaminoglycans/chemistry , Glycosaminoglycans/physiology , Proteoglycans/chemistry , Proteoglycans/physiology , Animals , Cell Line, Tumor , Epigenesis, Genetic , Extracellular Matrix/metabolism , Glycosaminoglycans/genetics , Humans , Neoplasms/physiopathology , Neurodegenerative Diseases/physiopathology , Protein Domains , Proteoglycans/genetics , Signal Transduction/physiology
15.
J Biol Chem ; 293(43): 16940-16950, 2018 10 26.
Article in English | MEDLINE | ID: mdl-30049794

ABSTRACT

The extracellular matrix is a master regulator of tissue homeostasis in health and disease. Here we examined how the small, leucine-rich, extracellular matrix proteoglycan decorin regulates cardiomyocyte metabolism during fasting in vivo First, we validated in Dcn-/- mice that decorin plays an essential role in autophagy induced by fasting. High-throughput metabolomics analyses of cardiac tissue in Dcn-/- mice subjected to fasting revealed striking differences in the hexosamine biosynthetic pathway resulting in aberrant cardiac O-ß-N-acetylglycosylation as compared with WT mice. Functionally, Dcn-/- mice maintained cardiac function at a level comparable with nonfasted animals whereas fasted WT mice showed reduced ejection fraction. Collectively, our results suggest that reduced sensing of nutrient deprivation in the absence of decorin preempts functional adjustments of cardiac output associated with metabolic reprogramming.


Subject(s)
Autophagy , Decorin/physiology , Extracellular Matrix/metabolism , Metabolome , Myocytes, Cardiac/pathology , Nutrients/metabolism , Animals , Cellular Reprogramming , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/metabolism
16.
J Biol Chem ; 293(31): 12137-12148, 2018 08 03.
Article in English | MEDLINE | ID: mdl-29921586

ABSTRACT

Regulation of autophagy by proteolytically cleaved fragments of heparan sulfate proteoglycans is a novel and current research focus in tumor biology. Endorepellin is the C-terminal angiostatic fragment of the heparan sulfate proteoglycan perlecan and induces autophagy in endothelial cells. To further investigate this property, we used NanoString, a digital PCR platform for measuring pre-defined transcripts in biological samples to analyze a custom subset of 95 autophagy-related genes in human umbilical vein endothelial cells treated with ultrapure human recombinant endorepellin. We discovered an endorepellin-evoked pro-autophagic and pro-mitophagic gene expression signatures, which included two coordinately up-regulated mitochondrial-associated genes encoding the E3 ubiquitin protein ligase Parkin and the tumor suppressor mitostatin. Induction of both proteins required the tyrosine kinase activity of vascular endothelial growth factor receptor 2 (VEGFR2). Furthermore, we discovered that endorepellin evoked mitochondrial depolarization in endothelial cells via a specific interaction between its two proximal LG1/2 domains and VEGFR2. We also found that following loss of membrane potential, mitostatin and parkin interact and that mitostatin associates with the established Parkin receptor mitofusin-2. In conclusion, we have identified a critical role for endorepellin in remodeling the autophagic transcriptome and influencing mitochondrial homeostasis.


Subject(s)
Autophagy , Heparan Sulfate Proteoglycans/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Mitophagy , Peptide Fragments/metabolism , Carrier Proteins , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Heparan Sulfate Proteoglycans/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Peptide Fragments/genetics , Protein Binding , Signal Transduction , Transcriptome , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
17.
Kidney Int ; 95(3): 540-562, 2019 03.
Article in English | MEDLINE | ID: mdl-30712922

ABSTRACT

Biglycan, a small leucine-rich proteoglycan, acts as a danger signal and is classically thought to promote macrophage recruitment via Toll-like receptors (TLR) 2 and 4. We have recently shown that biglycan signaling through TLR 2/4 and the CD14 co-receptor regulates inflammation, suggesting that TLR co-receptors may determine whether biglycan-TLR signaling is pro- or anti-inflammatory. Here, we sought to identify other co-receptors and characterize their impact on biglycan-TLR signaling. We found a marked increase in the number of autophagic macrophages in mice stably overexpressing soluble biglycan. In vitro, stimulation of murine macrophages with biglycan triggered autophagosome formation and enhanced the flux of autophagy markers. Soluble biglycan also promoted autophagy in human peripheral blood macrophages. Using macrophages from mice lacking TLR2 and/or TLR4, CD14, or CD44, we demonstrated that the pro-autophagy signal required TLR4 interaction with CD44, a receptor involved in adhesion, migration, lymphocyte activation, and angiogenesis. In vivo, transient overexpression of circulating biglycan at the onset of renal ischemia/reperfusion injury (IRI) enhanced M1 macrophage recruitment into the kidneys of Cd44+/+ and Cd44-/- mice but not Cd14-/- mice. The biglycan-CD44 interaction increased M1 autophagy and the number of renal M2 macrophages and reduced tubular damage following IRI. Thus, CD44 is a novel signaling co-receptor for biglycan, an interaction that is required for TLR4-CD44-dependent pro-autophagic activity in macrophages. Interfering with the interaction between biglycan and specific TLR co-receptors could represent a promising therapeutic intervention to curtail kidney inflammation and damage.


Subject(s)
Acute Kidney Injury/immunology , Biglycan/metabolism , Hyaluronan Receptors/metabolism , Macrophages/immunology , Reperfusion Injury/immunology , Acute Kidney Injury/pathology , Animals , Autophagosomes/immunology , Autophagosomes/metabolism , Autophagy/immunology , Biglycan/genetics , Biglycan/immunology , Cells, Cultured , Disease Models, Animal , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/immunology , Kidney Tubules/blood supply , Kidney Tubules/immunology , Kidney Tubules/pathology , Macrophage Activation , Mice , Mice, Knockout , Primary Cell Culture , Reperfusion Injury/pathology , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
18.
Biochem Soc Trans ; 47(5): 1543-1555, 2019 10 31.
Article in English | MEDLINE | ID: mdl-31652436

ABSTRACT

The extracellular matrix is a network of secreted macromolecules that provides a harmonious meshwork for the growth and homeostatic development of organisms. It conveys multiple signaling cascades affecting specific surface receptors that impact cell behavior. During cancer growth, this bioactive meshwork is remodeled and enriched in newly formed blood vessels, which provide nutrients and oxygen to the growing tumor cells. Remodeling of the tumor microenvironment leads to the formation of bioactive fragments that may have a distinct function from their parent molecules, and the balance among these factors directly influence cell viability and metastatic progression. Indeed, the matrix acts as a gatekeeper by regulating the access of cancer cells to nutrients. Here, we will critically evaluate the role of selected matrix constituents in regulating tumor angiogenesis and provide up-to-date information concerning their primary mechanisms of action.


Subject(s)
Neoplasms/blood supply , Neoplasms/metabolism , Neovascularization, Pathologic , Animals , Glycoproteins/chemistry , Glycoproteins/physiology , Humans , Proteoglycans/physiology , Thrombospondins/physiology , Tumor Microenvironment
19.
Dev Dyn ; 247(5): 679-685, 2018 05.
Article in English | MEDLINE | ID: mdl-29368411

ABSTRACT

BACKGROUND: Prostate ductal branching morphogenesis involves a complex spatiotemporal regulation of cellular proliferation and remodeling of the extracellular matrix (ECM) around the developing ducts. Decorin (Dcn) is a small leucine-rich proteoglycan known to sequester several growth factors and to act as a tumor suppressor in prostate cancer. RESULTS: Dcn expression in the developing prostate paralleled branching morphogenesis and was dynamically regulated by androgen and Hedgehog (Hh) signaling. DCN colocalized with collagen in the periductal stroma and acellular interstitium. Exogenous DCN decreased epithelial proliferation in ex vivo organ cultures of developing prostate, whereas genetic ablation of Dcn resulted in increased epithelial proliferation in the developing prostate. CONCLUSIONS: Dcn expression and localization in the developing prostate is consistent with a primary role in organizing collagen around the developing ducts. Regulation of Dcn expression appears to be complex, involving both androgen and Hh signaling. The growth inhibitory effect of Dcn suggests a unique linkage between a structural proteoglycan and epithelial growth regulation. This may serve to coordinate two elements of the morphogenetic process: ductal growth and organization of the collagen matrix around the nascent duct. Developmental Dynamics 247:679-685, 2018. © 2018 Wiley Periodicals, Inc.


Subject(s)
Decorin/metabolism , Prostate/metabolism , Signal Transduction/physiology , Animals , Decorin/genetics , Female , Male , Mice , Morphogenesis/physiology , Organ Culture Techniques , Organogenesis/physiology , Prostate/embryology , Prostate/growth & development
20.
J Biol Chem ; 292(39): 16211-16220, 2017 09 29.
Article in English | MEDLINE | ID: mdl-28798237

ABSTRACT

Macroautophagy is a fundamental and evolutionarily conserved catabolic process that eradicates damaged and aging macromolecules and organelles in eukaryotic cells. Decorin, an archetypical small leucine-rich proteoglycan, initiates a protracted autophagic program downstream of VEGF receptor 2 (VEGFR2) signaling that requires paternally expressed gene 3 (PEG3). We have discovered that PEG3 is an upstream transcriptional regulator of transcription factor EB (TFEB), a master transcription factor of lysosomal biogenesis, for decorin-evoked endothelial cell autophagy. We found a functional requirement of PEG3 for TFEB transcriptional induction and nuclear translocation in human umbilical vein endothelial and PAER2 cells. Mechanistically, inhibiting VEGFR2 or AMP-activated protein kinase (AMPK), a major decorin-activated energy sensor kinase, prevented decorin-evoked TFEB induction and nuclear localization. In conclusion, our findings indicate a non-canonical (nutrient- and energy-independent) mechanism underlying the pro-autophagic bioactivity of decorin via PEG3 and TFEB.


Subject(s)
Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/agonists , Decorin/metabolism , Endothelium, Vascular/metabolism , Kruppel-Like Transcription Factors/metabolism , Receptors, Growth Factor/agonists , Signal Transduction , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Aorta/cytology , Aorta/drug effects , Aorta/metabolism , Autophagy/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cells, Cultured , Decorin/genetics , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Gene Expression Regulation/drug effects , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Kruppel-Like Transcription Factors/chemistry , Kruppel-Like Transcription Factors/genetics , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Kinase Inhibitors/pharmacology , RNA Interference , Receptors, Growth Factor/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Sus scrofa , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL